Safety and immunogenicity of SpikoGen®, an Advax-CpG55.2-adjuvanted SARS-CoV-2 spike protein vaccine: a phase 2 randomized placebo-controlled trial in both seropositive and seronegative populations

Payam Tabarsi, Nassim Anjidani, Ramin Shahpari, Masoud Mardani, Araz Sabzvari, Babak Yazdani, Khashayar Roshanzamir, Behnam Bayatani, Ali Taheri, Nikolai Petrovsky, Lei Li, Saghar Barati, Payam Tabarsi, Nassim Anjidani, Ramin Shahpari, Masoud Mardani, Araz Sabzvari, Babak Yazdani, Khashayar Roshanzamir, Behnam Bayatani, Ali Taheri, Nikolai Petrovsky, Lei Li, Saghar Barati

Abstract

Objective: We aimed to investigate the immunogenicity and safety of SpikoGen®, a subunit COVID-19 vaccine composed of a recombinant prefusion-stabilized SARS-CoV-2 spike protein combined with the Advax-CpG55.2™ adjuvant, in seronegative and seropositive populations as primary vaccination.

Methods: This randomized, placebo-controlled, double-blind phase 2 trial was conducted on 400 participants randomized 3:1 to receive two doses of 25 μg of SpikoGen® 3 weeks apart or the placebo. The primary safety outcomes were the incidence of solicited adverse events up to 7 days after each dose and unsolicited adverse events up to 28 days after the second dose. The primary immunogenicity outcomes were seroconversion against the S1 protein and the geometric mean concentration of S1 antibodies by days 21 and 35.

Results: The SpikoGen® vaccine was well tolerated and no serious adverse events were recorded. The most common solicited adverse events were injection site pain and fatigue, largely graded as mild and transient. By day 35 (2 weeks post second dose), the seroconversion rate against S1 was 63.55 (95% CI: 57.81-69.01) in the SpikoGen® group versus 7.23 (95% CI: 2.7-15.07) in the placebo group. The geometric mean concentration of S1 antibodies was 29.12 (95% CI: 24.32-34.87) in the SpikoGen® group versus 5.53 (95% CI: 4.39-6.97) in the placebo group. Previously infected seropositive volunteers showed a large SARS-CoV-2 humoral response after a single SpikoGen® dose.

Discussion: SpikoGen® had an acceptable safety profile and induced promising humoral and cellular immune responses against SARS-CoV-2.

Keywords: Advax-CpG; COVID-19; Phase 2; SARS-CoV-2; SpikoGen; Subunit protein vaccine.

Copyright © 2022 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved.

Figures

Fig. 1
Fig. 1
Flowchart of screening, randomization, and analysis of the participants. There was an individual who received an incorrect injection during the study. This volunteer in the SpikoGen® group received a single dose of placebo by error and did not receive the vaccine. The participant was included in the placebo safety population. This participant also developed COVID-19 and therefore was excluded from the Per Protocol analysis.
Fig. 2
Fig. 2
Solicited local and systemic AEs. The percentage of participants in each group (SpikoGen®, Placebo) with Adverse Events (AEs) according to the maximum Food and Drug Administration (FDA) toxicity grading scale during the 7 days after each vaccination is plotted for solicited local (Panel A) and systemic (Panel B) adverse events. There was no grade 4 (life-threatening) event.
Fig. 3
Fig. 3
Shown are geometric mean concentrations (GMCs) in the per-protocol set for S1 IgG responses (Panel A), receptor binding domain IgG responses (Panel B), and surrogate virus neutralization test responses (Panel C) at day 21 (day of the second injection) and day 35 (14 days after the second injection). Antibody values below the lower limit of quantification (LLOQ) were replaced by 0.5 × LLOQ. The 95% CI was calculated based on the t-distribution of the log-transformed values for GMC levels, then back transformed to the original scale for presentation. For each group, geometric means are depicted above the scatterplot.
Fig. 4
Fig. 4
T cell response after stimulation with spike peptide pools. Shown are interferon-γ release after stimulation with AG1 (Panel A), and AG2 (Panel B), fold rise in Interferon-γ concentrations after stimulation with AG1 (Panel C) and AG2 (Panel D). Data are presented as median and interquartile range.

References

    1. Nascimento I.P., Leite L.C.C. Recombinant vaccines and the development of new vaccine strategies. Braz J Med Biol Res. 2012;45:1102–1111.
    1. Jeyanathan M., Afkhami S., Smaill F., Miller M.S., Lichty B.D., Xing Z. Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol. 2020;20:615–632.
    1. Richmond P., Hatchuel L., Dong M., Ma B., Hu B., Smolenov I., et al. Safety and immunogenicity of S-Trimer (SCB-2019), a protein subunit vaccine candidate for COVID-19 in healthy adults: a phase 1, randomised, double-blind, placebo-controlled trial. Lancet. 2021;397:682–694.
    1. Liang Z., Zhu H., Wang X., Jing B., Li Z., Xia X., et al. Adjuvants for coronavirus vaccines. Front Immunol. 2020;11:589833.
    1. Heath P.T., Galiza E.P., Baxter D.N., Boffito M., Browne D., Burns F., et al. Safety and efficacy of NVX-CoV2373 Covid-19 vaccine. N Engl J Med. 2021;385:117–183.
    1. Keech C., Albert G., Cho I., Robertson A., Reed P., Neal S., et al. Phase 1-2 trial of a SARS-CoV-2 recombinant spike protein nanoparticle vaccine. N Engl J Med. 2020;383:2320–2332.
    1. Bravo L., Smolenov I., Han H.H., Li P., Hosain R., Rockhold F., et al. Efficacy of the adjuvanted subunit protein COVID-19 vaccine, SCB-2019: a phase 2 and 3 multicentre, double-blind, randomised, placebo-controlled trial. Lancet. 2022;399:461–472.
    1. Gordon D.L., Sajkov D., Woodman R.J., Honda-Okubo Y., Cox M.M.J., Heinzel S., et al. Randomized clinical trial of immunogenicity and safety of a recombinant H1N1/2009 pandemic influenza vaccine containing Advax™ polysaccharide adjuvant. Vaccine. 2012;30:5407–5416.
    1. Gordon D., Kelley P., Heinzel S., Cooper P., Petrovsky N. Immunogenicity and safety of Advax™, a novel polysaccharide adjuvant based on delta inulin, when formulated with hepatitis B surface antigen: a randomized controlled Phase 1 study. Vaccine. 2014;32:6469–6477.
    1. Honda-Okubo Y., Saade F., Petrovsky N. Advax™, a polysaccharide adjuvant derived from delta inulin, provides improved influenza vaccine protection through broad-based enhancement of adaptive immune responses. Vaccine. 2012;30:5373–5381.
    1. Li L., Honda-Okubo Y., Huang Y., Jang H., Carlock M.A., Baldwin J., et al. Immunisation of ferrets and mice with recombinant SARS-CoV-2 spike protein formulated with Advax-SM adjuvant protects against COVID-19 infection. Vaccine. 2021;39:5940–5953.
    1. Honda-Okubo Y., Ong C.H., Petrovsky N. Advax delta inulin adjuvant overcomes immune immaturity in neonatal mice thereby allowing single-dose influenza vaccine protection. Vaccine. 2015;33:4892–4900.
    1. Sakala I.G., Honda-Okubo Y., Li L., Baldwin J., Petrovsky N. A M2 protein-based universal influenza vaccine containing Advax-SM adjuvant provides newborn protection via maternal or neonatal immunization. Vaccine. 2021;39:5162–5172.
    1. Honda-Okubo Y., Kolpe A., Li L., Petrovsky N. A single immunization with inactivated H1N1 influenza vaccine formulated with delta inulin adjuvant (Advax™) overcomes pregnancy-associated immune suppression and enhances passive neonatal protection. Vaccine. 2014;32:4651–4659.
    1. Eichinger K.M., Kosanovich J.L., Lipp M.A., Perkins T.N., Petrovsky N., Marshall C., et al. Maternal immunization with adjuvanted RSV prefusion F protein effectively protects offspring from RSV challenge and alters innate and T cell immunity. Vaccine. 2020;38:7885–7891.
    1. Honda-Okubo Y., Barnard D., Ong C.H., Peng B.-H., Tseng C.-T.K., Petrovsky N. Severe acute respiratory syndrome-associated coronavirus vaccines formulated with delta inulin adjuvants provide enhanced protection while ameliorating lung eosinophilic immunopathology. J Virol. 2015;89:2995–2997.
    1. Adney D.R., Wang L., van Doremalen N., Shi W., Zhang Y., Kong W.-P., et al. Efficacy of an adjuvanted Middle East Respiratory Syndrome Coronavirus spike protein vaccine in dromedary camels and alpacas. Viruses. 2019;11:212.
    1. Popmihajlov Z., Pang L., Brown E., Joshi A., Su S.-C., Kaplan S.S., et al. A post hoc analysis utilizing the FDA toxicity grading scale to assess injection site adverse events following immunization with the live attenuated Zoster Vaccine (ZVL) Hum Vaccin Immunother. 2018;14:2916–2920.
    1. FDA . 2020. Development and licensure of vaccines to prevent COVID-19. Available from:
    1. Dunkle L.M., Kotloff K.L., Gay C.L., Anez G., Adelglass J.M., Hernandez A.Q.B., et al. Efficacy and safety of NVX-CoV2373 in adults in the United States and Mexico. N Engl J Med. 2022;386:531–543.
    1. Krammer F., Srivastava K., Alshammary H., Amoako A.A., Awawda M.H., Beach K.F., et al. Antibody responses in seropositive persons after a single dose of SARS-CoV-2 mRNA vaccine. N Engl J Med. 2021;384:1372–1374.
    1. Jaganathan S., Stieber F., Rao S.N., Nikolayevskyy V., Manissero D., Allen N., et al. Preliminary evaluation of QuantiFERON SARS-CoV-2 and QIAreach Anti-SARS-CoV-2 total test in recently vaccinated individuals. Infect Dis Ther. 2021;10:2765–2776.
    1. Logunov D.Y., Dolzhikova I.V., Zubkova O.V., Tukhvatulin A.I., Shcheblyakov D.V., Dzharullaeva A.S., et al. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet. 2020;396:887–897.
    1. Li H., Monslow M.A., Freed D.C., Chang D., Li F., Gindy M., et al. Novel adjuvants enhance immune responses elicited by a replication-defective human cytomegalovirus vaccine in nonhuman primates. Vaccine. 2021;39:7446–7456.
    1. Chu R.S., Targoni O.S., Krieg A.M., Lehmann P.V., Harding C.V. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Th1) immunity. J Exp Med. 1997;186:1623–1631.
    1. Murad Y.M., Clay T.M. CpG oligodeoxynucleotides as TLR9 agonists: therapeutic applications in cancer. BioDrugs. 2009;23:361–375.

Source: PubMed

Подписаться